Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Medicine (Baltimore) ; 103(15): e37411, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38608087

RESUMO

BACKGROUND: Colonoscopy is a commonly performed gastroenterological procedure in patients associated with anxiety and pain. Various approaches have been used to provide sedation and analgesia during colonoscopy, including patient-controlled analgesia and sedation (PCAS). This study aims to evaluate the feasibility and efficiency of PCAS administered with propofol and remifentanil for colonoscopy. METHODS: This randomized controlled trial was performed in an authorized and approved endoscopy center. A total of 80 outpatients were recruited for the colonoscopy studies. Patients were randomly allocated into PCAS and total intravenous anesthesia (TIVA) groups. In the PCAS group, the dose of 0.1 ml/kg/min of the mixture was injected after an initial bolus of 3 ml mixture (1 ml containing 3 mg of propofol and 10 µg of remifentanil). Each 1 ml of bolus was delivered with a lockout time of 1 min. In the TIVA group, patients were administered fentanyl 1 µg/kg, midazolam 0.02 mg/kg, and propofol (dosage titrated). Cardiorespiratory parameters and auditory evoked response index were continuously monitored during the procedure. The recovery from anesthesia was assessed using the Aldrete scale and the Observer's Assessment of Alertness/Sedation Scale. The Visual Analogue Scale was used to assess the satisfaction of patients and endoscopists. RESULTS: No statistical differences were observed in the Visual Analogue Scale scores of the patients (9.58 vs 9.50) and the endoscopist (9.43 vs 9.30). A significant decline in the mean arterial blood pressure, heart rate, and auditory evoked response index parameters was recorded in the TIVA group (P < 0.05). The recovery time was significantly shorter in the PCAS group than in the TIVA group (P = 0.00). CONCLUSION: The combination of remifentanil and propofol could provide sufficient analgesia, better hemodynamic stability, lighter sedation, and faster recovery in the PCAS group of patients compared with the TIVA group.


Assuntos
Agnosia , Propofol , Humanos , Remifentanil , Midazolam , Analgesia Controlada pelo Paciente , Fentanila , Anestesia Intravenosa , Anestesia Geral , Colonoscopia , Dor
2.
Cell Stem Cell ; 31(4): 484-498.e5, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38458193

RESUMO

Alginate-encapsulated hepatocyte transplantation is a promising strategy to treat liver failure. However, its clinical application was impeded by the lack of primary human hepatocytes and difficulty in controlling their quality. We previously reported proliferating human hepatocytes (ProliHHs). Here, quality-controlled ProliHHs were produced in mass and engineered as liver organoids to improve their maturity. Encapsulated ProliHHs liver organoids (eLO) were intraperitoneally transplanted to treat liver failure animals. Notably, eLO treatment increased the survival of mice with post-hepatectomy liver failure (PHLF) and ameliorated hyperammonemia and hypoglycemia by providing liver functions. Additionally, eLO treatment protected the gut from PHLF-augmented permeability and normalized the increased serum endotoxin and inflammatory response, which facilitated liver regeneration. The therapeutic effect of eLO was additionally proved in acetaminophen-induced liver failure. Furthermore, we performed assessments of toxicity and biodistribution, demonstrating that eLO had no adverse effects on animals and remained non-tumorigenic.


Assuntos
Falência Hepática Aguda , Falência Hepática , Humanos , Camundongos , Animais , Falência Hepática Aguda/terapia , Falência Hepática Aguda/induzido quimicamente , Distribuição Tecidual , Células Cultivadas , Hepatócitos , Fígado , Falência Hepática/terapia , Falência Hepática/metabolismo , Organoides/metabolismo
3.
J Psychiatr Res ; 163: 180-194, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37216772

RESUMO

BACKGROUND: Posttraumatic stress disorder (PTSD), a psychiatric disorder caused by stressful events, is characterized by long-lasting fear memory. The nucleus accumbens shell (NAcS) is a key brain region that regulates fear-associated behavior. Small-conductance calcium-activated potassium channels (SK channels) play a key role in regulating the excitability of NAcS medium spiny neurons (MSNs) but their mechanisms of action in fear freezing are unclear. METHOD: We established an animal model of traumatic memory using conditioned fear freezing paradigm, and investigated the alterations in SK channels of NAc MSNs subsequent to fear conditioning in mice. We then utilized an adeno-associated virus (AAV) transfection system to overexpress the SK3 subunit and explore the function of the NAcS MSNs SK3 channel in conditioned fear freezing. RESULTS: Fear conditioning activated NAcS MSNs with enhanced excitability and reduced the SK channel-mediated medium after-hyperpolarization (mAHP) amplitude. The expression of NAcS SK3 were also reduced time-dependently. The overexpression of NAcS SK3 impaired conditioned fear consolidation without affecting conditioned fear expression, and blocked fear conditioning-induced alterations in NAcS MSNs excitability and mAHP amplitude. Additionally, the amplitudes of mEPSC, AMPAR/NMDAR ratio, and membrane surface GluA1/A2 expression in NAcS MSNs was increased by fear conditioning and returned to normal levels upon SK3 overexpression, indicating that fear conditioning-induced decrease of SK3 expression caused postsynaptic excitation by facilitating AMPAR transmission to the membrane. CONCLUSION: These findings show that the NAcS MSNs SK3 channel plays a critical role in conditioned fear consolidation and that it may influence PTSD pathogenesis, making it a potential therapeutic target against PTSD.


Assuntos
Transtornos Fóbicos , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Camundongos , Animais , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Núcleo Accumbens/metabolismo , Congelamento , Medo
4.
Cell Stem Cell ; 30(5): 617-631.e8, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37059100

RESUMO

Liver resection is the first-line treatment for primary liver cancers, providing the potential for a cure. However, concerns about post-hepatectomy liver failure (PHLF), a leading cause of death following extended liver resection, have restricted the population of eligible patients. Here, we engineered a clinical-grade bioartificial liver (BAL) device employing human-induced hepatocytes (hiHeps) manufactured under GMP conditions. In a porcine PHLF model, the hiHep-BAL treatment showed a remarkable survival benefit. On top of the supportive function, hiHep-BAL treatment restored functions, specifically ammonia detoxification, of the remnant liver and facilitated liver regeneration. Notably, an investigator-initiated study in seven patients with extended liver resection demonstrated that hiHep-BAL treatment was well tolerated and associated with improved liver function and liver regeneration, meeting the primary outcome of safety and feasibility. These encouraging results warrant further testing of hiHep-BAL for PHLF, the success of which would broaden the population of patients eligible for liver resection.


Assuntos
Falência Hepática , Fígado Artificial , Humanos , Animais , Suínos , Hepatócitos , Falência Hepática/cirurgia , Regeneração Hepática
5.
Environ Sci Pollut Res Int ; 29(56): 84300-84311, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35780272

RESUMO

Benzene is one of the most common occupational hazards in the working environment which was in the list of group 1 carcinogens. This study applied four occupational health risk assessment models: EPA model; MOM model of Singapore; the International Council on Mining and Metals (ICMM) model, and the Technical guide WS/T 777-2021 of China. The models assessed both non-carcinogenic and carcinogenic effects of benzene for 1629 employees in 50 factories in Jiangsu Province (China) who were exposed to benzene in the working environment and analysis the risk between industries by principal component analysis (PCA) method. The highest occupational health hazard of benzene among the five industries is petroleum processing industry, then followed by chemical products manufacturing industry, special equipment manufacturing industry, wood processing and products industry, and at last the pharmaceutical manufacturing industry. The population of abnormal routine blood parameters in the subjects was mostly in the "wood products industry" group, and the concentration of benzene in "wood products industry" group is the lowest in 5 groups. The industries with low exposure concentration have higher blood abnormality rates; this may be caused by the fact that blood damage is more secretive under low occupational health risk.


Assuntos
Exposição Ocupacional , Saúde Ocupacional , Humanos , Benzeno/análise , Exposição Ocupacional/análise , Projetos de Pesquisa , Indústrias , Medição de Risco/métodos , Carcinógenos/análise
6.
Hepatology ; 76(6): 1690-1705, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35229337

RESUMO

BACKGROUND AND AIMS: Hepatocyte transplantation has been demonstrated to be effective to treat liver metabolic disease and acute liver failure. Nevertheless, the shortage of donor hepatocytes restrained its application in clinics. To expand human hepatocytes at a large scale, several dedifferentiation-based protocols have been established, including proliferating human hepatocytes (ProliHH). However, the decreased transplantation efficiency of these cells after long-term expansion largely impedes their application. APPROACH AND RESULTS: We found that accompanied with dedifferentiation, long-term cultured ProliHH (lc-ProliHH) up-regulated a panel of chemokines and cytokines related to innate immunity, which were referred to as dedifferentiation-associated inflammatory factors (DAIF). DAIF elicited excessive macrophage responses, accounting for the elimination of lc-ProliHH specifically during engraftment. Two possible strategies to increase ProliHH transplantation were then characterized. Blockage of innate immune response by dexamethasone reverted the engraftment and repopulation of lc-ProliHH to a level comparable to primary hepatocytes, resulting in improved liver function and a better survival of fumarylacetoacetate hydrolase-deficient mice. Alternatively, rematuration of lc-ProliHH as organoids reduced the expression of DAIF and led to markedly improved engraftment. CONCLUSIONS: These results revealed that lc-ProliHH triggers exacerbated macrophage activation by DAIF and provided potential solutions for clinical transplantation of lc-ProliHH.


Assuntos
Hepatócitos , Fígado , Humanos , Camundongos , Animais , Hepatócitos/metabolismo , Fígado/metabolismo , Citocinas/metabolismo , Quimiocinas/metabolismo , Macrófagos/metabolismo
7.
Stem Cell Res Ther ; 13(1): 114, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35313986

RESUMO

BACKGROUND: The progression of Biliary Atresia (BA) is associated with the number of reactive ductular cells (RDCs) whose heterogeneity in origin and evolution in humans remains unknown. SOX9-positive liver progenitor-like cells (LPLCs) have been shown to participate in RDCs and new hepatocyte formation during cholestatic liver regeneration in an animal model, which implies the possibility that hepatocyte-reprogrammed LPLCs could be a source of RDCs in BA. The present study aimed to elucidate the characteristics of SOX9-positive LPLCs in BA for exploring new possible therapeutic targets by manipulating the bi-differentiation process of LPLCs to prevent disease progression. METHODS: Twenty-eight patients, including 24 patients with BA and 4 patients with Congenital Choledochal Cyst as the control group, were retrospectively recruited. Liver biopsy samples were classified histologically using a 4-point scale based on fibrosis severity. LPLCs were detected by SOX9 and HNF4A double positive staining. Single immunohistochemistry, double immunohistochemistry, and multiple immunofluorescence staining were used to determine the different cell types and characteristics of LPLCs. RESULTS: The prognostic predictors of BA, namely total bile acid (TBA), RDCs, and fibrosis, were correlated to the emergence of LPLCs. SOX9 and HNF4A double-positive LPLCs co-stained rarely with relevant markers of portal hepatic progenitor cells (portal-HPCs), including CK19, CK7, EPCAM, PROM1 (CD133), TROP2, and AFP. Under cholestasis conditions, LPLCs acquired superior proliferation and anti-senescence ability among hepatocytes. Moreover, LPLCs arranged as a pseudo-rosette structure appeared from the periportal parenchyma to the portal region, which implied the differentiation from hepatocyte-reprogrammed LPLCs to RDCs with the progression of cholestasis. CONCLUSIONS: LPLCs are associated with disease progression and prognostic factors of BA. The bipotent characteristics of LPLCs are different from those of portal-HPCs. As cholestasis progresses, LPLCs appear to gain superior proliferation and anti-senescence ability and continually differentiate to RDCs.


Assuntos
Atresia Biliar , Colestase , Fatores de Transcrição SOX9 , Atresia Biliar/complicações , Atresia Biliar/metabolismo , Atresia Biliar/patologia , Colestase/complicações , Colestase/patologia , Humanos , Fígado/metabolismo , Regeneração Hepática , Estudos Retrospectivos , Fatores de Transcrição SOX9/genética
8.
Xenotransplantation ; 28(4): e12702, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34145650

RESUMO

BACKGROUND: The human-to-rat hematopoietic stem cell transplantation (HSCT) model is rare, unlike its human-to-mouse counterpart. The rat models are desired, especially in areas of physiology, toxicology, and pharmacology. In addition to lymphocytes, macrophages are also considered to be important for xenotransplantation. We generated a rat xenotransplantation model to prove the role of macrophages as a xenotransplantation barrier. METHODS: Immunodeficiency in SRG rats, which are Sprague-Dawley (SD) rats lacking Rag2 and Il2rg, was confirmed by flow cytometry and spleen immunostaining. Human umbilical cord blood was collected after scheduled cesarean section at the University of Tsukuba Hospital. Cord blood mononuclear cells (CB-MNCs) were transplanted into the SRG rats administered several injections of clodronate liposome (CL), which cause macrophage depletion. Survival of human cells was observed by flow cytometry. Rat macrophage phagocytosis assay was performed to check the species-specific effects of rat macrophages on injected human/rat blood cells. RESULTS: SRG rats were deficient in T/B/NK cells. Without CL pretreatment, human CB-MNCs were removed from SRG rats within 7 hours after transplantation. The rats pretreated with CL could survive after transplantation. Prolonged survival for more than 4 weeks was observed only following a one-time CL injection. Rat macrophages had a species-specific potential for the phagocytosis of human blood cells in vivo. CONCLUSION: In human-to-rat HSCT, the short period of early macrophage control, leading to macrophage immunotolerance, is important for engraftment. The generated model can be useful for the creation of future xenotransplantation models or other clinical research.


Assuntos
Cesárea , Células-Tronco Hematopoéticas , Animais , Feminino , Humanos , Macrófagos , Camundongos , Camundongos SCID , Gravidez , Ratos , Ratos Sprague-Dawley , Transplante Heterólogo
9.
Cell Rep ; 33(10): 108455, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33296648

RESUMO

The ever-increasing therapeutic and pharmaceutical demand for liver cells calls for systems that enable mass production of hepatic cells. Here we describe a large-scale suspension system that uses human endoderm stem cells (hEnSCs) as precursors to generate functional and transplantable hepatocytes (E-heps) or cholangiocytes (E-chos). hEnSC-derived hepatic populations are characterized by single-cell transcriptomic analyses and compared with hESC-derived counterparts, in-vitro-maintained or -expanded primary hepatocytes and adult cells, which reveals that hepatic differentiation of hEnSCs recapitulates in vivo development and that the heterogeneities of the resultant populations can be manipulated by regulating the EGF and MAPK signaling pathways. Functional assessments demonstrate that E-heps and E-chos possess properties comparable with adult counterparts and that, when transplanted intraperitoneally, encapsulated E-heps were able to rescue rats with acute liver failure. Our study lays the foundation for cell-based therapeutic agents and in vitro applications for liver diseases.


Assuntos
Técnicas de Cultura de Células/métodos , Endoderma/citologia , Hepatócitos/citologia , Células-Tronco Embrionárias Humanas/citologia , Ductos Biliares/citologia , Ductos Biliares/metabolismo , Diferenciação Celular/fisiologia , Endoderma/metabolismo , Endoderma/transplante , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Hepatócitos/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/transplante , Humanos , Fígado/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante
10.
Methods Mol Biol ; 1905: 93-101, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30536093

RESUMO

Primary hepatocytes are widely used in regenerative medicine, drug metabolism analysis, and in vitro drug screens. To overcome the shortage of liver donors, several strategies, such as differentiation of pluripotent stem cells and transdifferentiation from somatic cells, were developed to generate hepatocytes from alternative sources. Here, we describe in detail lenti-virus-based procedure for direct conversion of human fibroblasts to hepatocytes (hiHep cells) in vitro. A detailed protocol for preparation of human fibroblasts from scar tissues is also provided. Based on this protocol, FOXA3, HNF1A, and HNF4A are introduced into SV40-large-T-antigen-expressing human scar fibroblasts by lenti-virus. It usually takes about 5-7 days to get epithelial hiHep colonies. SV40-large-T-antigen-expressing hiHep (hiHepLT) cells are proliferative and can be expanded to a large number for potential uses.


Assuntos
Técnicas de Cultura de Células/métodos , Cicatriz/patologia , Fibroblastos/citologia , Hepatócitos/citologia , Lentivirus/genética , Antígenos Transformantes de Poliomavirus/genética , Linhagem da Célula , Proliferação de Células , Transdiferenciação Celular , Reprogramação Celular , Cicatriz/genética , Cicatriz/metabolismo , Fibroblastos/metabolismo , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 3-gama Nuclear de Hepatócito/genética , Fator 3-gama Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Hepatócitos/metabolismo , Humanos , Vírus 40 dos Símios/genética
11.
Cell Stem Cell ; 23(6): 806-819.e4, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30416071

RESUMO

Transplantation of human hepatocytes (HHs) holds significant potential for treating liver diseases. However, the supply of transplantable HHs is severely constrained by limited donor availability and compromised capacity for in vitro expansion. In response to chronic injury, some HHs are reprogrammed into proliferative cells that express both hepatocyte and progenitor markers, suggesting exploitable strategies for expanding HHs in vitro. Here, we report defined medium conditions that allow 10,000-fold expansion of HHs. These proliferating HHs are bi-phenotypic, partially retaining hepatic features while gaining expression of progenitor-associated genes. Importantly, these cells engraft into injured mouse liver at a level comparable to primary HHs, and they undergo maturation following transplantation in vivo or differentiation in vitro. Thus, this study provides a protocol that enables large-scale expansion of transplantable HHs, which could be further developed for modeling and treating human liver disease.


Assuntos
Hepatócitos/citologia , Fígado/citologia , Animais , Contagem de Células , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID
12.
Stem Cell Reports ; 10(5): 1522-1536, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29606616

RESUMO

Disorders of the biliary epithelium, known as cholangiopathies, cause severe and irreversible liver diseases. The limited accessibility of bile duct precludes modeling of several cholangiocyte-mediated diseases. Therefore, novel approaches for obtaining functional cholangiocytes with high purity are needed. Previous work has shown that the combination of Hnf1ß and Foxa3 could directly convert mouse fibroblasts into bipotential hepatic stem cell-like cells, termed iHepSCs. However, the efficiency of converting fibroblasts into iHepSCs is low, and these iHepSCs exhibit extremely low differentiation potential into cholangiocytes, thus hindering the translation of iHepSCs to the clinic. Here, we describe that the expression of Hnf1α and Foxa3 dramatically facilitates the robust generation of iHepSCs. Notably, prolonged in vitro culture of Hnf1α- and Foxa3-derived iHepSCs induces a Notch signaling-mediated secondary conversion into cholangiocyte progenitor-like cells that display dramatically enhanced differentiation capacity into mature cholangiocytes. Our study provides a robust two-step approach for obtaining cholangiocyte progenitor-like cells using defined factors.


Assuntos
Sistema Biliar/citologia , Diferenciação Celular , Fibroblastos/citologia , Células-Tronco/citologia , Animais , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 3-gama Nuclear de Hepatócito/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Fígado/citologia , Camundongos Endogâmicos C57BL , Receptores Notch/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Transcrição Gênica
13.
J Biol Chem ; 293(18): 6883-6892, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29507093

RESUMO

Hereditary tyrosinemia type I (HTI) is a metabolic genetic disorder caused by mutation of fumarylacetoacetate hydrolase (FAH). Because of the accumulation of toxic metabolites, HTI causes severe liver cirrhosis, liver failure, and even hepatocellular carcinoma. HTI is an ideal model for gene therapy, and several strategies have been shown to ameliorate HTI symptoms in animal models. Although CRISPR/Cas9-mediated genome editing is able to correct the Fah mutation in mouse models, WT Cas9 induces numerous undesired mutations that have raised safety concerns for clinical applications. To develop a new method for gene correction with high fidelity, we generated a Fah mutant rat model to investigate whether Cas9 nickase (Cas9n)-mediated genome editing can efficiently correct the Fah First, we confirmed that Cas9n rarely induces indels in both on-target and off-target sites in cell lines. Using WT Cas9 as a positive control, we delivered Cas9n and the repair donor template/single guide (sg)RNA through adenoviral vectors into HTI rats. Analyses of the initial genome editing efficiency indicated that only WT Cas9 but not Cas9n causes indels at the on-target site in the liver tissue. After receiving either Cas9n or WT Cas9-mediated gene correction therapy, HTI rats gained weight steadily and survived. Fah-expressing hepatocytes occupied over 95% of the liver tissue 9 months after the treatment. Moreover, CRISPR/Cas9-mediated gene therapy prevented the progression of liver cirrhosis, a phenotype that could not be recapitulated in the HTI mouse model. These results strongly suggest that Cas9n-mediated genome editing is a valuable and safe gene therapy strategy for this genetic disease.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Desoxirribonuclease I/metabolismo , Edição de Genes , Terapia Genética/métodos , Tirosinemias/genética , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Células HEK293 , Hepatócitos/citologia , Humanos , Hidrolases/genética , Mutação INDEL , Cirrose Hepática/etiologia , Cirrose Hepática/prevenção & controle , Masculino , Ratos , Tirosinemias/complicações , Tirosinemias/imunologia , Tirosinemias/terapia
14.
Stem Cell Reports ; 9(6): 1813-1824, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29173899

RESUMO

Hepatocyte-like cells (HLCs) can be generated through directed differentiation or transdifferentiation. Employing two strategies, we generated induced pluripotent stem cell (iPSC)-HLCs and hiHeps from the same donor cell line. Both types of HLCs clustered distinctly from each other during gene expression profiling. In particular, differences existed in gene expression for phase II drug metabolism and lipid accumulation, underpinned by H3K27 acetylation status in iPSC-HLCs and hiHeps. While distinct phenotypes were achieved in vitro, both types of HLCs demonstrated similar phenotypes following transplantation into Fah-deficient mice. In conclusion, functional HLCs can be obtained from the same donor using two strategies. Global gene expression defined the differences between those populations in vitro. Importantly, both HLCs displayed partial but markedly improved hepatic function following transplantation in vivo, demonstrating plasticity and the potential for cell-based modeling in the dish and cell-based therapy in the future.


Assuntos
Diferenciação Celular/genética , Terapia Baseada em Transplante de Células e Tecidos , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Linhagem Celular , Epigênese Genética/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hepatócitos/metabolismo , Hepatócitos/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Camundongos , Doadores de Tecidos
15.
Sci Rep ; 6: 31460, 2016 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-27510266

RESUMO

Hereditary tyrosinemia type I (HT1) is caused by a deficiency in the enzyme fumarylacetoacetate hydrolase (Fah). Fah-deficient mice and pigs are phenotypically analogous to human HT1, but do not recapitulate all the chronic features of the human disorder, especially liver fibrosis and cirrhosis. Rats as an important model organism for biomedical research have many advantages over other animal models. Genome engineering in rats is limited till the availability of new gene editing technologies. Using the recently developed CRISPR/Cas9 technique, we generated Fah(-/-) rats. The Fah(-/-) rats faithfully represented major phenotypic and biochemical manifestations of human HT1, including hypertyrosinemia, liver failure, and renal tubular damage. More importantly, the Fah(-/-) rats developed remarkable liver fibrosis and cirrhosis, which have not been observed in Fah mutant mice or pigs. Transplantation of wild-type hepatocytes rescued the Fah(-/-) rats from impending death. Moreover, the highly efficient repopulation of hepatocytes in Fah(-/-) livers prevented the progression of liver fibrosis to cirrhosis and in turn restored liver architecture. These results indicate that Fah(-/-) rats may be used as an animal model of HT1 with liver cirrhosis. Furthermore, Fah(-/-) rats may be used as a tool in studying hepatocyte transplantation and a bioreactor for the expansion of hepatocytes.


Assuntos
Hepatócitos/transplante , Hidrolases/genética , Cirrose Hepática/prevenção & controle , Tirosinemias/patologia , Animais , Sistemas CRISPR-Cas , Células Cultivadas , Modelos Animais de Doenças , Hepatócitos/citologia , Humanos , Fígado/citologia , Fígado/patologia , Cirrose Hepática/patologia , Ratos , Tirosinemias/complicações , Tirosinemias/genética
16.
Cell Res ; 26(2): 206-16, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26768767

RESUMO

Acute liver failure (ALF) is a life-threatening illness. The extracorporeal cell-based bioartificial liver (BAL) system could bridge liver transplantation and facilitate liver regeneration for ALF patients by providing metabolic detoxification and synthetic functions. Previous BAL systems, based on hepatoma cells and non-human hepatocytes, achieved limited clinical advances, largely due to poor hepatic functions, cumbersome preparation or safety concerns of these cells. We previously generated human functional hepatocytes by lineage conversion (hiHeps). Here, by improving functional maturity of hiHeps and producing hiHeps at clinical scales (3 billion cells), we developed a hiHep-based BAL system (hiHep-BAL). In a porcine ALF model, hiHep-BAL treatment restored liver functions, corrected blood levels of ammonia and bilirubin, and prolonged survival. Importantly, human albumin and α-1-antitrypsin were detectable in hiHep-BAL-treated ALF pigs. Moreover, hiHep-BAL treatment led to attenuated liver damage, resolved inflammation and enhanced liver regeneration. Our findings indicate a promising clinical application of the hiHep-BAL system.


Assuntos
Hepatócitos/metabolismo , Hepatócitos/fisiologia , Falência Hepática Aguda/metabolismo , Falência Hepática Aguda/fisiopatologia , Albuminas/metabolismo , Amônia/metabolismo , Animais , Bilirrubina/metabolismo , Linhagem Celular , Humanos , Fígado Artificial , Suínos
17.
Cell Stem Cell ; 14(3): 370-84, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24582927

RESUMO

The generation of large numbers of functional human hepatocytes for cell-based approaches to liver disease is an important and unmet goal. Direct reprogramming of fibroblasts to hepatic lineages could offer a solution to this problem but so far has only been achieved with mouse cells. Here, we generated human induced hepatocytes (hiHeps) from fibroblasts by lentiviral expression of FOXA3, HNF1A, and HNF4A. hiHeps express hepatic gene programs, can be expanded in vitro, and display functions characteristic of mature hepatocytes, including cytochrome P450 enzyme activity and biliary drug clearance. Upon transplantation into mice with concanavalin-A-induced acute liver failure and fatal metabolic liver disease due to fumarylacetoacetate dehydrolase (Fah) deficiency, hiHeps restore the liver function and prolong survival. Collectively, our results demonstrate successful lineage conversion of nonhepatic human cells into mature hepatocytes with potential for biomedical and pharmaceutical applications.


Assuntos
Reprogramação Celular , Fibroblastos/citologia , Hepatócitos/citologia , Adulto , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Sistema Biliar/metabolismo , Diferenciação Celular/genética , Linhagem da Célula , Proliferação de Células , Concanavalina A , Feto/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/transplante , Humanos , Hidrolases/deficiência , Hidrolases/metabolismo , Inativação Metabólica , Falência Hepática/patologia , Falência Hepática/terapia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/citologia , Células-Tronco/metabolismo
18.
J Cell Biochem ; 114(2): 256-65, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22948752

RESUMO

One of the essential features of stem cells is their cellular plasticity to differentiate into daughter cells with defined functions. Recently, induction of pluripotent stem cells from somatic cells by defined transcription factors led to the focus on cellular plasticity of terminally differentiated cells. This approach is adopted by other studies to demonstrate the cell fate conversion between different lineages of terminally differentiated cells. We and others showed that induced hepatocyte-like (iHep) cells are directly converted from mouse fibroblasts by overexpression of liver-enriched transcription factors. iHep cells as well as pluripotent stem cell- or mesenchymal stem cell-derived hepatocyte-like cells provide potential cell sources for disease modeling, transplantation, and tissue engineering independent of donor organs. Here, we review the latest advances in generating hepatocyte-like cells and summarize general criteria for evaluating these cells. In addition, we propose a possible role of the p19(Arf) /p53 pathway in cell fate maintenance, which apparently limits the formation of induced pluripotent stem (iPS) cells and iHep cells.


Assuntos
Diferenciação Celular/genética , Fibroblastos , Hepatócitos , Células-Tronco Pluripotentes Induzidas , Animais , Linhagem da Célula , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Células-Tronco Pluripotentes/citologia , Transdução de Sinais , Fatores de Transcrição/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
19.
Artigo em Chinês | MEDLINE | ID: mdl-22468307

RESUMO

OBJECTIVE: To explore the relevance between chronic benzene poisoning and ABO blood type. METHODS: 1014 benzene-exposed workers chosen from Shanghai and 4196 non-benzene-exposed workers chosen from Yangpu district were accepted the ABO blood type identification, and two groups of workers compared with the Han population of Shanghai and China for the ABO blood type distribution; the 71 cases of chronic benzene poisoning were compared with the group of benzene-exposed workers for the ABO blood type distribution. RESULTS: There were no significant differences of the ABO blood type distribution among the Han population of China, the Han population of Shanghai and the group of non-benzene-exposed workers (x(2)=7.95, P>0.05). The A type blood distribution frequency in the group of chronic benzene poisoning patients was 42.25%, significantly higher than the group of benzene-exposed workers (29.48%), and there was statistically significant difference (x(2)=5.11,P<0.05). The B type blood distribution frequency in the group of chronic benzene poisoning patients was 12.68% , significantly lower than the group of benzene-exposed workers (25.15%), and there was statistically significant difference (x(2)=5.61, P0.05). CONCLUSION: The people with A type blood are susceptible to chronic benzene poisoning, however, the people with B type blood are not susceptible to chronic benzene poisoning.


Assuntos
Sistema ABO de Grupos Sanguíneos , Benzeno/intoxicação , Exposição Ocupacional , Sistema ABO de Grupos Sanguíneos/genética , Sistema ABO de Grupos Sanguíneos/metabolismo , China , Suscetibilidade a Doenças , Feminino , Humanos , Masculino
20.
Mol Cell Biol ; 30(24): 5621-35, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20937768

RESUMO

Resent studies have identified Pygopus as a core component of the ß-catenin/T-cell factor (TCF)/lymphoid-enhancing factor 1 (LEF) transcriptional activation complex required for the expression of canonical Wg/Wnt target genes in Drosophila. However, the biochemical involvement of mammalian Pygopus proteins in ß-catenin/TCF/LEF gene activation remains controversial. In this study, we perform a series of molecular/biochemical experiments to demonstrate that Pygo2 associates with histone-modifying enzymatic complexes, specifically the MLL2 histone methyltransferase (HMT) and STAGA histone acetyltransferase (HAT) complexes, to facilitate their interaction with ß-catenin and to augment Wnt1-induced, TCF/LEF-dependent transcriptional activation in breast cancer cells. We identify a critical domain in Pygo2 encompassing the first 47 amino acids that mediates its HMT/HAT interaction. We further demonstrate the importance of this domain in Pygo2's ability to transcriptionally activate both artificial and endogenous Wnt target genes and to expand breast cancer stem-like cells in culture. This work now links mechanistically Pygo2's role in histone modification to its enhancement of the Wnt-dependent transcriptional program and cancer stem-like cell expansion.


Assuntos
Neoplasias da Mama , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/fisiologia , Fatores de Transcrição de p300-CBP/metabolismo , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Feminino , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/citologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Fatores de Transcrição de p300-CBP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA